CN112480122B - Tetrahydroimidazo [1,2-a ] pyrazine compound, composition, preparation method and application thereof - Google Patents

Tetrahydroimidazo [1,2-a ] pyrazine compound, composition, preparation method and application thereof Download PDF

Info

Publication number
CN112480122B
CN112480122B CN202011327191.4A CN202011327191A CN112480122B CN 112480122 B CN112480122 B CN 112480122B CN 202011327191 A CN202011327191 A CN 202011327191A CN 112480122 B CN112480122 B CN 112480122B
Authority
CN
China
Prior art keywords
compound
phenyl
tert
dihydroimidazo
piperazin
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
CN202011327191.4A
Other languages
Chinese (zh)
Other versions
CN112480122A (en
Inventor
熊小峰
张小雷
葛阳
石硕
邓俊杰
刘璐
陈新滋
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Sun Yat Sen University
Original Assignee
Sun Yat Sen University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Sun Yat Sen University filed Critical Sun Yat Sen University
Priority to CN202011327191.4A priority Critical patent/CN112480122B/en
Publication of CN112480122A publication Critical patent/CN112480122A/en
Application granted granted Critical
Publication of CN112480122B publication Critical patent/CN112480122B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Oncology (AREA)
  • Hematology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention discloses a tetrahydroimidazo [1,2-a ]]Pyrazine compounds, compositions, and methods of preparation and use thereof. The compound has a structure shown in formula (I), has G protein resistant activity superior to BIM-46174, and has no toxicity to normal cells; and the preparation is simple and the cost is low.

Description

Tetrahydroimidazo [1,2-a ] pyrazine compound, composition, preparation method and application thereof
Technical Field
The invention belongs to the technical field of medicines, and particularly relates to a tetrahydroimidazo [1,2-a ] pyrazine compound, a composition, a preparation method and an application thereof.
Background
G protein-coupled receptors (GPCRs) are the largest family of cell surface receptors, over 800 known GPCRs have been identified in the human genome, and GPCRs are widely expressed in vivo and are involved in a large number of physiological and pathological processes. GPCRs mediate a variety of physiological functions by coupling to the intracellular heterotrimeric guanine nucleotide binding protein (G protein), passing a number of extracellular signals through the cell membrane. GPCRs are currently the most common class of drug targets, with about 25% of drugs approved for marketing by the FDA in the united states targeting GPCRs, suggesting that GPCR-G protein signaling pathways play an important role in drug development.
The heterotrimeric G protein is used as a molecular switch in the signal transduction process of a G protein-coupled receptor (GPCR), transmits information from the outside of the cell to the inside of the cell, and regulates the growth and development process of organisms through a series of cascade reactions. The G protein is located inside the cell membrane and is a heterotrimer composed of α, β, and γ subunits, with different subunits performing different biological functions. When a GPCR is activated, a signal can be transmitted to the downstream G protein through a conformational change, resulting in activation of the G protein. When the G protein is in the inactive state, the G α subunit binds Guanosine Diphosphate (GDP) and forms a stable trimer with the dimeric G β γ subunit. When the G protein is activated, G.alpha.changes from the inactivated state G.alpha.GDP bound to GDP to the activated state G.alpha.GTP bound to Guanosine Triphosphate (GTP), and then G.alpha.and G.beta.gamma.are separated and the respective signaling is initiated. The activated G protein will initiate further signaling, activating a number of "second messenger" systems and initiating a range of important physiological activities. The signal is terminated by the G protein signal regulatory protein (RGS) binding to G.alpha.subunit to play the role of GAP (GTPase activating protein) to hydrolyze GTP into GDP, and then the G.alpha.subunit binding to GDP is recombined with G.beta.gamma subunit to form G.alpha.gamma heterotrimer, and the G protein returns to the inactive state.
G proteins can be classified as G.alpha.based on the homology of the G protein's alpha subunitq/11、Gαi、GαsAnd G.alpha.12/13Four subfamilies. G alphasCan increase the catalytic activity of downstream Adenosine Cyclase (AC) and lead to the increase of the cyclic adenosine monophosphate (cAMP) level of a second messenger, and then activate Protein Kinase A (PKA) to trigger a series of downstream signal transmission processes. And G alphasIn contrast, G.alpha.iThe activation of (b) inhibits the activity of AC, resulting in a decrease in cAMP levels. G alpha12/13Is capable of activating small GTPases. G alphaq/11Is capable of activating phospholipase C beta (PLC beta), which is capable of hydrolyzing 4, 5-diphosphatidyinositol (PIP2) and producing two distinct "second messengers," Diacylglycerol (DAG) and inositol triphosphate (IP3), wherein DAG is capable of activating Protein Kinase C (PKC), and IP3 is capable of activating ligand-gated Ca on the endoplasmic reticulum2+Ion channels and promote Ca2+Ion release to the cytoplasm, and the generation of DAG and IP3 will trigger physiological phenomena such as platelet aggregation, smooth muscle contraction, exocytosis and the like. In addition to normal physiological processes, G protein is an important mediator in a variety of tumors (e.g., uveal melanoma, prostate)Cancer, pancreatic cancer, leukemia, glioblastoma) and inflammation (e.g., asthma). The G protein has become one of the important targets for research and development of antitumor drugs.
The G protein small molecule inhibitor has good prospect for treating malignant tumor and inflammatory diseases. FR900359 and YM-254890 as highly selective Gaq/11Inhibitors, IC thereof5032nM and 95nM, respectively, have been used as tool molecules for studying uveal melanoma and asthma (Onken, m., et al. sci. signal.,2018,11, eaao 6852; Matthey, m., et al. sci. trans. med.,2017,9, eaag 2288), but there is a great limitation in disease treatment due to great difficulty in synthesis. EC for inhibition of G protein by Suramin50240nM (Freismuth, M., et al. mol. Pharmacol.,1996,49, 602-611.). BIM-46174 is a G protein inhibitor (US7034024B1, WO0134203A1, EP1430934A1) developed by French SOD CONSEILS RECH APPLIC, and its dimer BIM-46187 is directed to G.alpha.q/11The selectivity of (Ayou ub, M., et al J. biol. chem.,2009,284, 29136-29145; Schmitz, A., et al chem. biol.,2014,21, 890-902) is high. Other compounds, such as 0990(Appleton, k., et al bioorg.med.chem.,2014,22,3423-iProtein selectivity; m119 and Gallein are highly selective G.beta.gamma.inhibitors, the IC of which50Is 200nM (Bonacci, T., et al. science,2006,312, 443-. The small molecule inhibitor has the problems of high toxicity, poor structural stability, low antitumor activity and the like, so that the subsequent development and research of the small molecule inhibitor are limited.
In view of the urgent need for cancer treatment, there is a need in the art to develop new drugs with better efficacy.
Disclosure of Invention
The invention aims to overcome the defects in the prior art and provides a tetrahydroimidazo [1,2-a ] pyrazine compound or pharmaceutically acceptable salt thereof, wherein the compound has good antitumor activity.
The present invention also provides a pharmaceutical composition of the above tetrahydroimidazo [1,2-a ] pyrazines compound or a pharmaceutically acceptable salt thereof.
The invention further aims to provide the tetrahydroimidazo [1,2-a ] pyrazine compound or pharmaceutically acceptable salt thereof, or application of the composition.
The purpose of the invention is realized by the following technical scheme: a tetrahydroimidazo [1,2-a ] pyrazine compound or a pharmaceutically acceptable salt thereof, wherein the chemical structure of the tetrahydroimidazo [1,2-a ] pyrazine compound is represented by the formula (I):
Figure BDA0002794657540000021
wherein the content of the first and second substances,
x is selected from carbonyl or sulfonyl;
l is selected from-CH-or hydrogen;
R1selected from hydrogen or methyl;
R2selected from hydrogen, methyl, ethyl, isopropyl, hydroxyl, mercapto, carboxyl, cyano,
Figure BDA0002794657540000022
Figure BDA0002794657540000023
R3Selected from methyl, amino, hydroxyl,
Figure BDA0002794657540000024
R4Selected from methyl, isopropyl, and isopropyl,
Figure BDA0002794657540000025
As a specific embodiment of the invention, the compound represented by the general formula (I) has a structure shown in formulas I-1 to I-36:
Figure BDA0002794657540000031
the structural compound is a tetrahydroimidazo [1,2-a ] pyrazine compound, and the screening of the antitumor activity of the invention shows that part of the compounds have stronger capability of inhibiting the proliferation of uveal melanoma cells (92.1 and MP41), and part of the compounds show better anti-G protein activity than BIM-46174. As a molecule with novel structure, the compound has the potential of being developed into a novel high-efficiency G protein inhibitor, and has great application value in treating related tumor diseases, particularly uveal melanoma, prostatic cancer, acute myeloid leukemia, pancreatic cancer or glioblastoma.
The structures represented by the foregoing I-1 to I-36 have the following names, respectively:
(I-1) (S) -3-amino-4- ((S) -8- (cyclohexylmethyl) -2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) -4-oxobutanenitrile;
(I-2) (S) -2-amino-1- ((S) -8- (cyclohexylmethyl) -2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) -3-methylbutan-1-one;
(I-3) (S) -2-amino-1- ((S) -8- (cyclohexylmethyl) -2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) -3- (4-hydroxyphenyl) propan-1-one;
(I-4) (S) -2-amino-1- ((S) -8- (cyclohexylmethyl) -2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) -3- (1H-imidazol-4-yl) propan-1-one;
(I-5) (S) -2-amino-1- ((S) -8- (cyclohexylmethyl) -2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) propan-1-one;
(I-6) (S) -2-amino-1- ((S) -8- (cyclohexylmethyl) -2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) -4-methylpentane-1-one;
(I-7) (S) -2-amino-1- ((S) -8- (cyclohexylmethyl) -2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) -3-phenylpropan-1-one;
(I-8) (S) -3-amino-4- ((S) -8- (cyclohexylmethyl) -2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) -4-oxobutanoic acid;
(I-9)1- ((S) -4-amino-5- ((S) -8- (cyclohexylmethyl) -2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) -5-oxopentyl) guanidine;
(I-10) (2S,3R) -2-amino-1- ((S) -8- (cyclohexylmethyl) -2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) -3-methylpentane-1-one;
(I-11) (S) -4-amino-5- ((S) -8- (cyclohexylmethyl) -2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) -5-oxopentanoic acid;
(I-12) (S) -3-amino-4- ((S) -8- (cyclohexylmethyl) -2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) -4-oxobutanamide;
(I-13) (2S,3R) -2-amino-1- ((S) -8- (cyclohexylmethyl) -2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) -3-hydroxybutan-1-one;
(I-14) (S) -2, 6-diamino-1- ((S) -8- (cyclohexylmethyl) -2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) hexan-1-one;
(I-15) (S) -4-amino-5- ((S) -8- (cyclohexylmethyl) -2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) -5-oxovaleramide;
(I-16) (S) -2-amino-1- ((S) -8- (cyclohexylmethyl) -2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) -3- (1H-indol-3-yl) propan-1-one;
(I-17) (S) -2-amino-3-cyclohexyl-1- ((S) -8- (cyclohexylmethyl) -2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) propan-1-one;
(I-18) (S) -1- ((S) -8- (cyclohexylmethyl) -2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) -3-mercapto-2-methylaminopropane-1-one;
(I-19) (S) -1- ((S) -8- (cyclohexylmethyl) -2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) -2- (dimethylamino) -3-mercaptopropan-1-one;
(I-20) (S) -1- ((S) -8- (cyclohexylmethyl) -2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) -2- (diethylamino) -3-mercaptopropan-1-one;
(I-21) (S) -1- (8- (cyclohexylmethyl) -2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) -2-methylpropan-1-one;
(I-22)1- ((S) -8- (cyclohexylmethyl) -2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) -2-methylbutan-1-one;
(I-23) (2S,3R) -2-amino-1- ((S) -8-benzyl-2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) -3-methylpentane-1-one;
(I-24) (S) -2-amino-1- ((R) -8- (cyclohexylmethyl) -2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) -3- (4-hydroxyphenyl) propan-1-one;
(I-25) (S) -2-amino-1- ((R) -8-isobutyl-2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) -3-mercaptopropan-1-one;
(I-26) (S) -2-amino-1- ((S) -8- ((R) -sec-butyl) -2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) -3-mercaptopropan-1-one;
(I-27) (S) -2-amino-1- ((S) -8-isopropyl-2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) -3-mercaptopropan-1-one;
(I-28) (S) -2-amino-3-mercapto-1- ((S) -8-methyl-2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) propan-1-one;
(I-29) (S) -8- (cyclohexylmethyl) -7- (isopropylsulfonyl) -2-phenyl-5, 6,7, 8-tetrahydroimidazo [1,2-a ] piperazine;
(I-30) (S) -2-amino-1- (8- (cyclohexylmethyl) -2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) ethan-1-one;
(I-31) (S) -8- (cyclohexylmethyl) -7- (ethylsulfonyl) -2-phenyl-5, 6,7, 8-tetrahydroimidazo [1,2-a ] piperazine;
(I-32) (S) -8-benzyl-7- (isopropylsulfonyl) -2-phenyl-5, 6,7, 8-tetrahydroimidazo [1,2-a ] piperazine;
(I-33) (S) -2, 6-diamino-1- ((R) -8-isobutyl-2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) hexan-1-one;
(I-34) (S) -1- ((S) -8- (cyclohexylmethyl) -2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) -2-hydroxypropan-1-one;
(I-35) (R) -1- (8-isobutyl-2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) -2-methylpropan-1-one;
(I-36) (2S,3R) -2-amino-3-methyl-1- ((R) -8-methyl-2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) pentan-1-one.
The preparation method of the tetrahydroimidazo [1,2-a ] pyrazine compound comprises the following steps:
(1) dissolving a compound 1 with a structural formula shown as a formula 1 and an acid-binding agent A in an organic solvent A, adding a compound 2 with a structural formula shown as a formula 2 for reaction, and removing the organic solvent A after the reaction is finished; adding an organic solvent B for resuspension, adding ammonium acetate, carrying out reflux reaction, and removing the organic solvent B after the reaction is finished; adding water and an extracting agent for extraction, and taking an extracting agent layer; washing, dewatering and concentrating the obtained extractant layer to obtain a compound 3;
Figure BDA0002794657540000051
(2) dissolving the compound 3 and an acid-binding agent C in an organic solvent C, adding ethyl bromoacetate, cooling after reaction, and adding water to separate out a compound 4;
(3) dispersing compound 4 and catalyst D in organic solvent D, sealing, and charging H2Reacting, filtering, concentrating the obtained filtrate, and purifying to obtain a compound 5;
(4) dispersing the compound 5 and borane tetrahydrofuran complex solution in an organic solvent E, sealing, introducing argon, carrying out a first reflux reaction, and removing the organic solvent E after TLC monitoring reaction is finished; adding organic solvent F, refluxing for the second time, concentrating, and purifying to obtain compound 6
(5) Preparation of tetrahydroimidazo [1,2-a ] pyrazines by step (a) or by step (B):
(A) dissolving the compound 7 in an organic solvent G, and adding diisopropylcarbodiimide to carry out a first reaction; then adding a compound 6, carrying out a second reaction, and after the TLC monitoring reaction is finished, concentrating and purifying; adding CF to the obtained product under inert atmosphere3COOH and triisopropylsilane are reacted for the third time to obtain the tetrahydroimidazol [1,2-a ] with the structural formula shown in the formula (I)]Pyrazine compounds;
Figure BDA0002794657540000061
x is selected from carbonyl or sulfonyl; l is selected from-CH-or hydrogen; r1Selected from hydrogen or methyl;
R2selected from hydrogen, methyl, ethyl, isopropyl, hydroxyl, mercapto, carboxyl, cyano,
Figure BDA0002794657540000062
Figure BDA0002794657540000063
R3Selected from methyl, amino, hydroxyl,
Figure BDA0002794657540000064
R4Selected from methyl, isopropyl, and isopropyl,
Figure BDA0002794657540000065
(B) Dissolving the compound 8 and triethylamine in an organic solvent H, adding the compound 6, reacting, monitoring by TLC, concentrating and purifying to obtain the tetrahydroimidazo [1,2-a ] pyrazine compound with the structural formula shown in the formula (I).
The preparation method of the pharmaceutically acceptable salt is to react the tetrahydroimidazo [1,2-a ] pyrazine compound with organic acid or inorganic acid to obtain the pharmaceutically acceptable salt.
The compound 1 described in the step (1) is preferably (S) -2- (((benzyloxy) carbonyl) amino) -3-cyclohexylpropionic acid, N-benzyloxycarbonyl-L-phenylalanine, (R) -2- (((benzyloxy) carbonyl) amino) -3-cyclohexylpropionic acid, N-benzyloxycarbonyl-D-leucine, N-benzyloxycarbonyl-L-isoleucine, N-benzyloxycarbonyl-L-valine, N-benzyloxycarbonyl-L-alanine, N-benzyloxycarbonyl-D-leucine, N-benzyloxycarbonyl-D-alanine.
The acid-binding agent A in the step (1) is preferably Cs2CO3
The dosage of the acid-binding agent A is preferably the same molar weight as that of the compound 1.
The organic solvent A in step (1) is preferably DMF.
The organic solvent A is a reaction medium and is used for dissolving reaction substances; preferably, 30mL of organic solvent A is used per 15.3mmol of compound 1.
The reaction temperature in step (1) is room temperature.
The room temperature is 10-40 ℃; preferably 20-30 ℃; more preferably 24 to 28 ℃.
The reaction time in the step (1) is 10-30 min; preferably 20 min.
The organic solvent A is removed in the step (1) preferably by rotary evaporation.
The organic solvent B in the step (1) is preferably toluene.
The dosage of the ammonium acetate is preferably 8 to 10 times of the 1 molar weight of the compound; more preferably 9 times.
The temperature of the reflux reaction described in the step (1) depends on the boiling point of the organic solvent B used.
The time of the reflux reaction in the step (1) is preferably 1-3 h; preferably for 2 hours.
The organic solvent B is removed in the step (1) preferably by rotary evaporation.
The water in the step (1) is selectively added, and the use amount of the water is to increase the volume of the reactant, so that an extractant layer is obtained. If the extraction is repeated, water is added at the first extraction.
The extractant in the step (1) is preferably ethyl acetate.
The number of extractions described in step (1) is preferably 3.
The washing in step (1) is preferably carried out with a saturated brine.
The water removal in the step (1) is preferably performed by using anhydrous sodium sulfate.
The concentration in the step (1) is preferably performed by a reduced pressure rotary evaporation method.
In the step (2)The acid-binding agent C is preferably K2CO3
The dosage of the acid-binding agent C in the step (2) is preferably 1-2 times of the molar weight of the compound 3; more preferably 1.5 times.
The organic solvent C in step (2) is preferably DMF.
The organic solvent C in the step (2) is used for forming a solution reaction system and is a reaction medium; the amount of the compound is preferably less than 28.9mmol of the compound 3 and 50mL of the organic solvent C.
The reaction temperature in the step (2) is preferably 40-60 ℃; more preferably 50 deg.c.
The reaction time in the step (2) is preferably 1-3 h; more preferably 2 h.
The catalyst D described in step (3) is preferably Pd/C having a dry content of 10%.
The organic solvent D in the step (3) is preferably methanol.
The dosage of the organic solvent D is preferably 15mL of methanol per 7.36mmol of the compound 4.
The temperature of the reaction in the step (3) is room temperature.
The reaction time in the step (3) is preferably 6-10 h; more preferably 8 h.
The filtration described in step (3) is preferably by filtration through celite.
The compound 6 and the borane tetrahydrofuran complex in the step (4) are preferably mixed in a molar ratio of 1: 5-7; more preferably, the molar ratio of 1: 6 to prepare a mixture ratio.
The organic solvent E in the step (4) is preferably tetrahydrofuran.
The dosage of the organic solvent E is preferably 20mL per 9.7mmol of the compound 6.
The time for the first reflux reaction described in step (4) is preferably 8 hours.
The removal of the organic solvent E in step (4) is preferably carried out by rotary evaporation.
The time for the second reflux reaction in step (4) is preferably 2 hours.
The compound 7 in the step (A) is preferably N-t-butoxycarbonyl-L-asparagine, Boc-L-valine, Boc-O-t-butyl-L-tyrosine, N-t-butoxycarbonyl-N' -trityl-L-histidine, N-t-butoxycarbonyl-L-alanine, N-t-butoxycarbonyl-L-leucine, N-t-butoxycarbonyl-L-phenylalanine, t-butoxycarbonyl-L-aspartic acid-4-t-butyl ester, tri-t-butoxycarbonylarginine, N-t-butoxycarbonyl-L-isoleucine, N-t-butoxycarbonyl-L-glutamic acid-5-t-butyl ester, t-butoxycarbonyl-N-beta-trityl-L-asparagine, Boc-L-histidine, N-t-butyloxycarbonyl-L-alanine, N-t-butyloxycarbonyl-L-isoleucine, N-t-butoxycarbonyl-L-glutamic acid-5-t-butyl ester, N-beta-trityl-L-asparagine, or L-histidine, N-tert-butyloxycarbonyl-O-tert-butyl-L-threonine, (S) -2, 6-di-tert-butyloxycarbonylaminohexanoic acid, N-tert-butyloxycarbonyl-N '-trityl-L-glutamine, N-tert-butyloxycarbonyl-N' -tert-butyloxycarbonyl-L-tryptophan, (S) -2- ((tert-butoxycarbonyl) amino) -3-cyclohexylpropionic acid, N-Boc-N-methyl-S-trityl-L-cysteine, N-dimethyl-S-trityl-L-cysteine, N-diethyl-S-trityl-L-cysteine, isobutyric acid, and mixtures thereof, (S) -2-methylbutyric acid, N-tert-butoxycarbonyl-L-isoleucine, Boc-O-tert-butyl-L-tyrosine, N-tert-butoxycarbonyl-S-trityl-L-cysteine, N-tert-butoxycarbonylglycine, (S) -2, 6-di-tert-butoxycarbonylaminocaproic acid or O-tert-butoxycarbonyl-L-lactic acid.
The organic solvent G in step (A) is preferably dichloromethane.
The amount of the organic solvent G used in the step (A) is preferably 2mL per 0.43mmol of the compound (7).
The diisopropylcarbodiimide used in step (a) is used in excess relative to compound 7; preferably according to compound 7: diisopropylcarbodiimide in a molar ratio of 1: 1.01-2 proportion calculation; more preferably according to compound 7: diisopropylcarbodiimide in a molar ratio of 1: 1.5 calculating the mixture ratio.
The time for the first reaction described in step (A) is preferably 30 min.
The compound 6 is used in an excess amount relative to the compound 7 in the step (A); preferably according to compound 7: compound 6 in a molar ratio of 1: 1.01-1.2 proportion calculation; more preferably according to compound 7: compound 6 in a molar ratio of 1: 1.1 calculating the mixture ratio.
The time of the second reaction in the step (A) is preferably 6-16 h.
CF described in step (A)3The amount of COOH used is preferably 0.8 to 1.2mL of CF per 7mmol of the compound3Calculating COOH; 1mL of CF per 0.43mmol of compound 73COOH calculation.
The triisopropylsilane is used in an excess amount relative to compound 7 in step (A); preferably according to compound 7: triisopropylsilane in a molar ratio of 1: 1.01-1.2 proportion calculation; more preferably according to compound 7: triisopropylsilane in a molar ratio of 1: 1.1 calculating the mixture ratio.
The time for the third reaction described in step (A) is preferably 2 h.
The step (A) also comprises the steps of filtering, concentrating and purifying the reaction liquid obtained by the third reaction.
The purification is preferably by semi-preparative liquid phase purification.
The compound 8 described in step (B) is preferably isopropyl sulfonyl chloride or ethyl sulfonyl chloride.
The dosage of the triethylamine in the step (B) is 2-2.1 times of the molar weight of the compound 8.
The organic solvent H used in step (B) is preferably dichloromethane.
The amount of the organic solvent G used in the step (B) is preferably 2mL of the organic solvent H per 0.7mmol of the compound 8.
The molar amount of the compound 6 in the step (B) is equivalent to the molar amount of the compound 8.
The reaction in the step (B) is preferably carried out for 6-16 h at 35-45 ℃; more preferably, the reaction is carried out for 6 to 16 hours at 40 ℃.
The specific steps of purification described in steps (3), (4) and (5) are preferably as follows: performing silica gel column chromatography, and spin-drying.
The silica gel is preferably silica gel with the particle size of 200 and 300 meshes.
In another aspect, the present invention provides a pharmaceutical composition comprising an effective amount of a compound represented by the general formula (i) or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
The drug carrier is preferably liposome or nanoparticle.
The salts of the compounds of formula (I) are preferably pharmaceutically acceptable salts, because of their potential use in medicine. The compounds of the present invention are bases, wherein the desired salt form can be prepared by suitable methods known in the art, including treatment of the free base with a mineral acid, such as hydrochloric acid, hydrobromic acid, sulfuric acid, sulfurous acid, pyrosulfuric acid, nitric acid, phosphoric acid, and the like; or treating the free base with an organic acid, such as acetic acid, trifluoroacetic acid, maleic acid, succinic acid, mandelic acid, fumaric acid, malonic acid, pyruvic acid, oxalic acid, glycolic acid, salicylic acid, pyranosidyl acid (pyranosidy1 acid), such as glucuronic acid or galacturonic acid, an alpha-hydroxy acid, such as citric acid or tartaric acid, an amino acid, such as aspartic acid or glutamic acid, an aromatic acid, such as benzoic acid or cinnamic acid, a sulfonic acid, such as p-toluenesulfonic acid, methanesulfonic acid, ethanesulfonic acid, and the like. Examples of pharmaceutically acceptable salts include sulfate, pyrosulfate, bisulfate, sulfite, bisulfite, phosphate, chloride, bromide, iodide, acetate, propionate, caprate, caprylate, acrylate, formate, isobutyrate, hexanoate, heptanoate, propionate, oxalate, malonate, benzoates, chlorobenzoates, methylbenzoates, dinitrobenzoates, hydroxybenzoates, methoxybenzoates, phthalates, phenylacetates, phenylpropionates, phenylbutyrates (phenylbutyrates), citrates, lactates, gamma-hydroxybutyrates, hydroxyacetates, tartrates, mandelates and sulfonates, such as xylenesulfonates, methanesulfonates, propanesulfonates, naphthalene-1-sulfonate and naphthalene-2-sulfonate.
The pharmaceutical compositions of the invention will generally contain one compound of the invention. However, in some embodiments, the pharmaceutical compositions of the invention contain more than one compound of the invention. In addition, the pharmaceutical compositions of the present invention may optionally further comprise one or more other pharmaceutically active compounds.
The invention also provides the tetrahydroimidazo [1,2-a ] pyrazine compound or a pharmaceutically acceptable carrier thereof, and application of the pharmaceutical composition in inhibiting tumor proliferation by inhibiting G protein. Specifically, the application is mainly used for preparing a medicament for treating melanoma, breast cancer, lung cancer, pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, stomach cancer, intestinal cancer, head and neck cancer, anal cancer, extrahepatic and biliary tract cancer, bladder cancer, bone cancer, brain stem glioma, brain tumor, bronchial adenoma, burkitt's lymphoma, carcinoid tumor, unknown primary cancer, central nervous system lymph cancer, cervical cancer, children cancer, germ cell tumor, eye cancer, kidney cancer, laryngeal cancer, liver cancer, non-small cell lung cancer, rectal cancer, salivary gland carcinoma, sarcoma, small intestine cancer, soft tissue sarcoma, uterine sarcoma, testicular cancer, leukemia or blood lymphoma.
The invention provides an application of a compound shown in the specification or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of the invention in preparation of a G protein inhibitor.
The invention provides a compound shown in a general formula (I) or a pharmaceutically acceptable salt thereof, or application of a pharmaceutical composition in preparing a medicament for treating tumors. Preferably, the tumor is selected from uveal melanoma, prostate cancer, acute myeloid leukemia, pancreatic cancer or glioblastoma, further preferably uveal melanoma. More preferably, the use is primarily through inhibition of G-proteins.
Compared with the prior art, the invention has the following advantages and effects:
the anti-G protein activity of the tetrahydroimidazo [1,2-a ] pyrazine compound provided by the invention is superior to that of BIM-46174, and the compound has no toxicity to normal cells; and the preparation is simple and the cost is low.
Detailed Description
The present invention will be described in further detail with reference to examples, but the embodiments of the present invention are not limited thereto.
In the present example, the experimental method not specified for the specific conditions is generally performed under the conventional conditions or under the conditions recommended by the manufacturers of the raw materials or the commercial products. Reagents of specific sources are not indicated, and conventional reagents are purchased in the market.
EXAMPLE 1 preparation of target molecules
The thin layer chromatography silica gel plate is HSGF254 of tobacco yellow sea or GF254 of Qingdao, the silica-amine plate used in Thin Layer Chromatography (TLC) is 0.15-0.2 mm, and the thin layer chromatography separation and purification product is 0.4-0.5 mm.
The column chromatography is performed by using 200-mesh and 300-mesh silica gel, and can be purchased from Yangtze river friend silica gel development Co.
The raw materials used in the present invention are mainly available from companies such as Sahn's chemical technology (Shanghai) Co., Ltd, Shanghai Bide medicine science and technology Co., Ltd, national drug group chemical reagent Co., Ltd, and Aladdin chemical reagent Co., Ltd.
In the examples, the solution means an aqueous solution unless otherwise specified.
In the examples, the reaction temperature is, unless otherwise specified, from 20 ℃ to 30 ℃ at room temperature.
The technical scheme adopted by the invention is as follows:
Figure BDA0002794657540000101
synthetic routes, reagents and conditions for compound (i): a) cs2CO3N, N-Dimethylformamide (DMF), room temperature, 86-92%; b) ammonium acetate and toluene, refluxing, 70-85%; c) bromoacetic acid ethyl ester, K2CO3,DMF,50℃,90%;d)Pd/C,H262-80% of room temperature; e) BH3THF, reflux, 55-78%; f) DIC, DCM, room temperature, 75-90%; g) triethylamine and DCM at 40 ℃ and 60-85 percent; h) triisopropylsilane, CF3COOH, room temperature, 80-90%.
Synthesis of Compound 3:
Figure BDA0002794657540000102
taking compound 1(15.3mmol) and Cs2CO3(5.00g, 15.3mmol) in 30mL DMF, mix well and then slowlySlowly adding a compound 2(3.04g,15.3mmol), reacting at room temperature for 20min, after the reaction is finished, evaporating DMF, adding 30mL of methylbenzene for resuspending, taking ammonium acetate (10.6g,138mmol) in a reaction bottle, carrying out reflux reaction for 2h, evaporating methylbenzene, adding a proper amount of water, extracting with ethyl acetate (100mL multiplied by 3), combining ethyl acetate layers, washing with saturated salt water once, drying with anhydrous sodium sulfate, and carrying out reduced pressure evaporation to obtain a compound 3 of a reddish brown oily liquid.
Synthesis of Compound 4:
Figure BDA0002794657540000111
taking compound 3(28.9mmol) and K2CO3(6.02g,43.4mmol) in 50mL DMF, adding ethyl bromoacetate (5.07g,30.3mmol), heating to 50 ℃, reacting for 2 hours, cooling, adding 400mL water, precipitating off-white solid, filtering, drying to obtain off-white solid compound 4, and directly reacting in the next step without purification.
Synthesis of Compound 5:
Figure BDA0002794657540000112
taking compound 4(7.36mmol) and 10% Pd/C (0.36g) in 15mL of methanol, sealing, and filling with H2Reacting at room temperature for 8 hours, filtering with diatomite after the reaction is finished to obtain filtrate, concentrating, carrying out silica gel column chromatography, and spin-drying to obtain a white solid compound 5.
Synthesis of Compound 6:
Figure BDA0002794657540000113
and (2) taking the compound 5(9.70mmol) and 1mol/L borane tetrahydrofuran complex solution (58.2mL,58.2mmol) to be put in 20mL dry tetrahydrofuran, sealing, introducing argon, refluxing for 8 hours, evaporating to remove the tetrahydrofuran after TLC monitoring reaction is finished, adding methanol, refluxing for 2 hours, concentrating, carrying out silica gel column chromatography, and carrying out spin drying to obtain the compound 6 of light yellow oily liquid.
Synthesis of target (I):
Figure BDA0002794657540000114
taking the compound 7(0.43mmol), adding 2mL of Dichloromethane (DCM) into a reaction bottle, stirring at room temperature, adding Diisopropylcarbodiimide (DIC) (0.08g,0.65mmol) for reaction for 30 minutes, adding the compound 6(0.14g,0.47mmol), reacting at room temperature overnight, monitoring by TLC after the reaction is finished, concentrating, performing silica gel column chromatography, and spin-drying to obtain a white solid. Putting the white solid into a reaction bottle, sealing, introducing argon, adding 1mL of CF3COOH, stirring at room temperature, adding triisopropylsilane (0.11g,0.47mmol), reacting at room temperature for 2 hours, filtering after the reaction is finished, obtaining filtrate, concentrating, and purifying by semi-preparative liquid phase to obtain the target molecule (I).
Adding the compound 8(0.70mmol) and triethylamine (0.14g,1.41mmol) into 2mL DCM, adding the compound 6(0.21g,0.70mmol), heating to 40 ℃ for overnight reaction, monitoring the reaction by TLC, concentrating, performing silica gel column chromatography, and spin-drying to obtain the target molecule (I).
The target molecule was synthesized according to the above method, and the physicochemical data of the synthesized target molecule were as follows:
compound I-1: the name (S) -3-amino-4- ((S) -8- (cyclohexylmethyl) -2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) -4-oxobutanenitrile (compound 1 used was (S) -2- (((benzyloxy) carbonyl) amino) -3-cyclohexylpropionic acid, compound 7 was N-tert-butoxycarbonyl-L-asparagine);
1H NMR(400MHz,CD3OD):δ8.09(s,1H),7.97–7.86(m,2H),7.50–7.38(m,3H),5.96(t,J=6.8Hz,1H),4.77–4.71(m,1H),4.60–4.56(m,1H),4.32–4.15(m,2H),3.91–3.85(m,1H),3.01–2.88(m,2H),2.18–2.15(m,1H),1.99–1.96(m,2H),1.64–1.56(m,4H),1.44–0.91(m,6H);MS(ESI)m/z 392.2[M+H]+
compound I-2: the name (S) -2-amino-1- ((S) -8- (cyclohexylmethyl) -2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) -3-methylbutan-1-one (compound 1 used was (S) -2- (((benzyloxy) carbonyl) amino) -3-cyclohexylpropionic acid, compound 7 was Boc-L-valine);
1H NMR(400MHz,CD3OD)δ7.83(s,1H),7.74–7.70(m,2H),7.53–7.44(m,3H),6.11(dd,J=10.4,3.6Hz,1H),4.53(d,J=4.0Hz,1H),4.42(dd,J=12.8,4.0Hz,2H),4.29–4.22(m,1H),4.06–3.93(m,1H),2.31–2.22(m,2H),2.09–2.02(m,1H),1.82–1.64(m,5H),1.44–1.23(m,4H),1.18(d,J=6.8Hz,3H),1.10–1.02(m,2H),1.00(d,J=6.8Hz,3H);MS(ESI)m/z395.3[M+H]+.
compound I-3: the name (S) -2-amino-1- ((S) -8- (cyclohexylmethyl) -2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) -3- (4-hydroxyphenyl) propan-1-one (compound 1 used was (S) -2- (((benzyloxy) carbonyl) amino) -3-cyclohexylpropionic acid, compound 7 was Boc-O-tert-butyl-L-tyrosine);
1H NMR(400MHz,CD3OD)δ7.75–7.68(m,2H),7.59(s,1H),7.55–7.44(m,3H),7.03(d,J=8.4Hz,2H),6.59(d,J=8.8Hz,2H),6.06(dd,J=10.8,3.2Hz,1H),4.74(dd,J=9.2,6.4Hz,1H),4.20(dd,J=15.2,4.8Hz,1H),3.93(dd,J=12.8,4.0Hz,1H),3.87–3.74(m,1H),3.13–3.01(m,2H),2.51–2.44(m,1H),2.30(d,J=12.4Hz,1H),2.06–1.93(m,1H),1.77–1.61(m,5H),1.45–1.36(m,1H),1.33–1.16(m,3H),1.07–0.98(m,2H);MS(ESI)m/z 459.3[M+H]+
compound I-4: the name (S) -2-amino-1- ((S) -8- (cyclohexylmethyl) -2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) -3- (1H-imidazol-4-yl) propan-1-one (compound 1 used was (S) -2- (((benzyloxy) carbonyl) amino) -3-cyclohexylpropionic acid, compound 7 was N-tert-butoxycarbonyl-N' -trityl-L-histidine);
1H NMR(400MHz,CD3OD)δ8.87(d,J=1.2Hz,1H),7.86(s,1H),7.75–7.71(m,2H),7.55–7.44(m,4H),6.14(dd,J=10.8,3.2Hz,1H),5.05–5.00(m,1H),4.65(dd,J=15.6,4.0Hz,1H),4.44(dd,J=13.2,3.6Hz,1H),4.38–4.29(m,1H),4.14–3.99(m,1H),3.47(dd,J=16.0,4.4Hz,1H),3.30–3.20(m,1H),2.27(d,J=12.0Hz,1H),2.13–2.06(m,1H),1.83–1.63(m,5H),1.46–1.38(m,1H),1.35–1.19(m,3H),1.08–0.99(m,2H);MS(ESI)m/z 433.3[M+H]+.
compound I-5: the name (S) -2-amino-1- ((S) -8- (cyclohexylmethyl) -2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) propan-1-one (compound 1 used was (S) -2- (((benzyloxy) carbonyl) amino) -3-cyclohexylpropionic acid, compound 7 was N-tert-butoxycarbonyl-L-alanine);
1H NMR(400MHz,CD3OD)δ7.83(s,1H),7.73–7.69(m,2H),7.54–7.44(m,3H),6.09(dd,J=10.8,2.8Hz,1H),4.64(q,J=6.8Hz,1H),4.45–4.33(m,2H),4.32–4.23(m,1H),4.04–3.94(m,1H),2.26(d,J=12.0Hz,1H),2.10–2.03(m,1H),1.80–1.63(m,5H),1.54(d,J=6.8Hz,3H),1.40–1.20(m,4H),1.10–0.98(m,2H);MS(ESI)m/z 367.2[M+H]+
compound I-6: the name (S) -2-amino-1- ((S) -8- (cyclohexylmethyl) -2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) -4-methylpentane-1-one (compound 1 used was (S) -2- (((benzyloxy) carbonyl) amino) -3-cyclohexylpropionic acid, compound 7 was N-tert-butoxycarbonyl-L-leucine);
1H NMR(400MHz,CD3OD)δ7.82(s,1H),7.74–7.69(m,2H),7.53–7.43(m,3H),6.09(dd,J=10.8,3.2Hz,1H),4.66(dd,J=10.4,3.2Hz,1H),4.49–4.38(m,1H),4.33–4.24(m,2H),4.08–3.98(m,1H),2.25(d,J=12.0Hz,1H),2.11–2.01(m,1H),1.90–1.57(m,9H),1.47–1.15(m,5H),1.09(d,J=6.4Hz,3H),1.02(d,J=6.0Hz,3H);MS(ESI)m/z 409.2[M+H]+
compound I-7: the name (S) -2-amino-1- ((S) -8- (cyclohexylmethyl) -2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) -3-phenylpropan-1-one (compound 1 used was (S) -2- (((benzyloxy) carbonyl) amino) -3-cyclohexylpropionic acid, compound 7 was N-tert-butoxycarbonyl-L-phenylalanine);
1H NMR(400MHz,CD3OD)δ7.73–7.68(m,2H),7.56(s,1H),7.50(t,J=6.8Hz,2H),7.44(t,J=7.2Hz,1H),7.31–7.17(m,5H),6.03(dd,J=10.8,3.6Hz,1H),4.83–4.79(m,1H),4.16(dd,J=15.2,4.8Hz,1H),3.94–3.76(m,2H),3.18(d,J=7.2Hz,2H),2.42–2.35(m,1H),2.28(d,J=12.8Hz,1H),2.04–1.93(m,1H),1.78–1.64(m,5H),1.42–1.34(m,1H),1.29–1.20(m,3H),1.07–0.98(m,2H);MS(ESI)m/z 443.3[M+H]+
compound I-8: the name (S) -3-amino-4- ((S) -8- (cyclohexylmethyl) -2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) -4-oxobutanoic acid (compound 1 used was (S) -2- (((benzyloxy) carbonyl) amino) -3-cyclohexylpropionic acid, compound 7 was tert-butoxycarbonyl-L-aspartic acid-4-tert-butyl ester);
1H NMR(400MHz,CD3OD)δ7.81(s,1H),7.73–7.67(m,2H),7.52–7.43(m,3H),6.08(dd,J=11.2,3.2Hz,1H),4.93–4.92(m,1H),4.48(d,J=16.4Hz,1H),4.41–4.28(m,2H),4.05–3.97(m,1H),3.05(dd,J=18.0,6.0Hz,1H),2.87(dd,J=18.0,7.6Hz,1H),2.25(d,J=12.4Hz,1H),2.10–2.03(m,1H),1.82–1.66(m,5H),1.46–1.36(m,1H),1.34–1.20(m,3H),1.04(dd,J=22.4,10.8Hz,2H);MS(ESI)m/z 411.2[M+H]+
compound I-9: under the name 1- ((S) -4-amino-5- ((S) -8- (cyclohexylmethyl) -2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) -5-oxopentyl) guanidine (compound 1 used was (S) -2- (((benzyloxy) carbonyl) amino) -3-cyclohexylpropionic acid, compound 7 was tri-tert-butoxycarbonylarginine);
1H NMR(400MHz,CD3OD)δ7.84(s,1H),7.74–7.69(m,2H),7.54–7.44(m,3H),6.13(dd,J=10.8,2.8Hz,1H),4.70(dd,J=7.6,4.0Hz,1H),4.49–4.37(m,2H),4.30–4.25(m,1H),4.08–3.95(m,1H),3.25(t,J=6.8Hz,2H),2.26(d,J=12.0Hz,1H),2.11–1.96(m,2H),1.93–1.84(m,1H),1.81–1.61(m,7H),1.41–1.19(m,4H),1.08–0.95(m,2H);MS(ESI)m/z 452.3[M+H]+
compound I-10: the name (2S,3R) -2-amino-1- ((S) -8- (cyclohexylmethyl) -2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) -3-methylpentane-1-one (compound 1 used was (S) -2- (((benzyloxy) carbonyl) amino) -3-cyclohexylpropionic acid, compound 7 was N-tert-butoxycarbonyl-L-isoleucine);
1H NMR(400MHz,CD3OD)δ7.80(s,1H),7.71(d,J=6.8Hz,2H),7.52–7.43(m,3H),6.09(dd,J=10.0,3.6Hz,1H),4.53(d,J=4.0Hz,1H),4.44–4.38(m,2H),4.24–4.14(m,1H),4.06–3.95(m,1H),2.24(d,J=12.0Hz,1H),2.07–1.95(m,2H),1.82–1.65(m,5H),1.57–1.51(m,1H),1.46–1.38(m,1H),1.36–1.19(m,4H),1.16(d,J=7.2Hz,3H),1.08–1.00(m,2H),0.94(t,J=7.6Hz,3H);MS(ESI)m/z 409.3[M+H]+
compound I-11: the name (S) -4-amino-5- ((S) -8- (cyclohexylmethyl) -2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) -5-oxopentanoic acid (the compound 1(S) -2- (((benzyloxy) carbonyl) amino) -3-cyclohexylpropionic acid used, compound 7 is N-tert-butoxycarbonyl-L-glutamic acid-5-tert-butyl ester);
1H NMR(400MHz,CD3OD)δ7.85(s,1H),7.76–7.67(m,2H),7.56–7.43(m,3H),6.06(dd,J=11.6,3.2Hz,1H),4.74(dd,J=7.2,3.6Hz,1H),4.64–4.46(m,2H),4.40(dd,J=12.8,4.0Hz,1H),4.09–3.94(m,1H),2.68–2.53(m,2H),2.26(d,J=12.4Hz,1H),2.20–2.00(m,3H),1.81–1.61(m,5H),1.42–1.19(m,4H),1.04(dd,J=23.6,12.0Hz,2H);MS(ESI)m/z425.3[M+H]+.
compound I-12: the name (S) -3-amino-4- ((S) -8- (cyclohexylmethyl) -2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) -4-oxobutanamide (compound 1 used was (S) -2- (((benzyloxy) carbonyl) amino) -3-cyclohexylpropionic acid, compound 7 was tert-butoxycarbonyl-N-beta-trityl-L-asparagine);
1H NMR(400MHz,CD3OD)δ7.84(s,1H),7.74–7.69(m,2H),7.54–7.44(m,3H),6.12(dd,J=10.8,2.4Hz,1H),4.87–4.75(m,1H),4.50(d,J=15.6Hz,1H),4.40–4.33(m,2H),4.06–3.93(m,1H),2.99(dd,J=16.4,6.0Hz,1H),2.80(dd,J=16.8,7.2Hz,1H),2.27(d,J=12.0Hz,1H),2.13–2.01(m,1H),1.81–1.63(m,5H),1.45–1.35(m,1H),1.34–1.17(m,3H),1.04(dd,J=23.6,11.6Hz,2H);MS(ESI)m/z 410.3[M+H]+
compound I-13: the name (2S,3R) -2-amino-1- ((S) -8- (cyclohexylmethyl) -2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) -3-hydroxybutan-1-one (compound 1 used was (S) -2- ((((benzyloxy) carbonyl) amino) -3-cyclohexylpropionic acid, compound 7 was N-tert-butoxycarbonyl-O-tert-butyl-L-threonine);
1H NMR(400MHz,CD3OD)δ7.83(s,1H),7.74–7.69(m,2H),7.54–7.44(m,3H),6.14(d,J=8.4Hz,1H),4.64(d,J=3.2Hz,1H),4.50(d,J=14.8Hz,1H),4.42–4.31(m,2H),4.27–4.21(m,1H),4.07–3.86(m,1H),2.27(d,J=11.6Hz,1H),2.11–2.03(m,1H),1.81–1.64(m,5H),1.40–1.20(m,4H),1.30(d,J=6.0Hz,3H),1.08–0.99(m,2H);MS(ESI)m/z 397.3[M+H]+
compound I-14: the name (S) -2, 6-diamino-1- ((S) -8- (cyclohexylmethyl) -2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) hexan-1-one (compound 1 used was (S) -2- (((benzyloxy) carbonyl) amino) -3-cyclohexylpropionic acid, compound 7 was (S) -2, 6-di-tert-butoxycarbonylaminocaproic acid);
1H NMR(400MHz,CD3OD)δ7.82(s,1H),7.75–7.69(m,2H),7.55–7.42(m,3H),6.09(dd,J=10.8,3.2Hz,1H),4.66(dd,J=7.6,4.0Hz,1H),4.45–4.38(m,2H),4.30–4.20(m,1H),4.09–3.94(m,1H),2.93(t,J=8.0Hz,2H),2.25(d,J=12.0Hz,1H),2.06–2.02(m,1H),1.97–1.84(m,2H),1.81–1.63(m,7H),1.59–1.48(m,2H),1.40–1.19(m,4H),1.08–0.99(m,2H);MS(ESI)m/z 424.3[M+H]+
compound I-15: the name (S) -4-amino-5- ((S) -8- (cyclohexylmethyl) -2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) -5-oxopentanamide (compound 1 used was (S) -2- (((benzyloxy) carbonyl) amino) -3-cyclohexylpropionic acid, compound 7 was N-tert-butoxycarbonyl-N' -trityl-L-glutamine);
1H NMR(400MHz,CD3OD)δ7.85(d,J=4.8Hz,1H),7.74–7.68(m,2H),7.54–7.45(m,3H),6.19–6.04(m,1H),4.88–4.84(m,1H),4.72–4.55(m,1H),4.49–4.33(m,2H),4.07–3.88(m,1H),2.56–2.52(m,1H),2.44–2.32(m,1H),2.31–2.15(m,2H),2.15–1.99(m,2H),1.80–1.61(m,5H),1.39–1.18(m,4H),1.03(dd,J=22.0,10.0Hz,2H);MS(ESI)m/z 424.3[M+H]+
compound I-16: the title compound is (S) -2-amino-1- ((S) -8- (cyclohexylmethyl) -2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) -3- (1H-indol-3-yl) propan-1-one (compound 1 used is (S) -2- (((benzyloxy) carbonyl) amino) -3-cyclohexylpropionic acid, compound 7 is N-tert-butoxycarbonyl-N' -tert-butoxycarbonyl-L-tryptophan, 144599-95-1).
1H NMR(400MHz,CD3OD)δ7.69–7.59(m,2H),7.57–7.48(m,3H),7.37(d,J=7.2Hz,1H),7.29(s,1H),7.23(s,1H),7.19(dd,J=7.2,1.6Hz,1H),6.78–6.71(m,2H),5.82(dd,J=11.2,3.2Hz,1H),4.76(dd,J=9.6,6.0Hz,1H),4.14(dd,J=14.8,5.2Hz,1H),3.72–3.58(m,2H),3.46–3.33(m,2H),2.33(d,J=12.0Hz,1H),1.93–1.72(m,3H),1.69(d,J=9.6Hz,2H),1.62–1.52(m,2H),1.39–1.21(m,4H),1.04–0.95(m,2H);MS(ESI)m/z 482.3[M+H]+.
Compound I-17: the name was (S) -2-amino-3-cyclohexyl-1- ((S) -8- (cyclohexylmethyl) -2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) propan-1-one (compound 1 used was (S) -2- (((benzyloxy) carbonyl) amino) -3-cyclohexylpropionic acid, compound 7 was (S) -2- ((tert-butoxycarbonyl) amino) -3-cyclohexylpropionic acid).
1H NMR(400MHz,CD3OD)δ7.83(s,1H),7.75–7.69(m,2H),7.52–7.41(m,3H),6.06(dd,J=10.8,3.2Hz,1H),4.65(dd,J=10.4,3.2Hz,1H),4.50–4.38(m,1H),4.31–4.21(m,2H),4.09–3.97(m,1H),2.11–2.01(m,4H),1.90–1.57(m,10H),1.53–1.15(m,4H)1.38–1.20(m,4H),1.06–0.96(m,4H);MS(ESI)m/z 449.3[M+H]+
Compound I-18: the name was (S) -1- ((S) -8- (cyclohexylmethyl) -2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) -3-mercapto-2-methylaminopropane-1-one (compound 1 used was (S) -2- (((benzyloxy) carbonyl) amino) -3-cyclohexylpropionic acid, compound 7 was N-Boc-N-methyl-S-trityl-L-cysteine, 91292-54-5).
1H NMR(400MHz,CD3OD)δ7.85(s,1H),7.76–7.69(m,2H),7.54–7.44(m,3H),6.20(dd,J=11.2,3.2Hz,1H),4.83(t,J=5.6Hz,1H),4.50(dd,J=15.2,4.0Hz,1H),4.45–4.32(m,2H),4.09–3.97(m,1H),3.13(dd,J=15.2,6.0Hz,1H),3.07(dd,J=14.8,5.2Hz,1H),2.77(s,3H),2.30(d,J=12.0Hz,1H),2.16–2.05(m,1H),1.86–1.64(m,5H),1.34–1.18(m,4H),1.12–1.01(m,2H);MS(ESI)m/z 413.2[M+H]+.
Compound I-19: the name was (S) -1- ((S) -8- (cyclohexylmethyl) -2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) -2- (dimethylamino) -3-mercaptopropan-1-one (compound 1 used was (S) -2- (((benzyloxy) carbonyl) amino) -3-cyclohexylpropionic acid, compound 7 was N, N-dimethyl-S-trityl-L-cysteine, 1275955-00-4).
1H NMR(400MHz,CD3OD)δ7.82(s,1H),7.73–7.69(m,2H),7.53–7.43(m,3H),6.20(dd,J=11.6,3.2Hz,1H),4.89–4.86(m,1H),4.55(d,J=15.6Hz,1H),4.42–4.37(m,2H),4.11–3.98(m,1H),3.21–3.12(m,2H),2.98(s,6H),2.30(d,J=12.0Hz,1H),2.17–2.05(m,1H),1.86–1.67(m,5H),1.31–1.18(m,4H),1.12–1.03(m,2H);MS(ESI)m/z 427.3[M+H]+
Compound I-20: the name was (S) -1- ((S) -8- (cyclohexylmethyl) -2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) -2- (diethylamino) -3-mercaptopropan-1-one (compound 1 used was (S) -2- (((benzyloxy) carbonyl) amino) -3-cyclohexylpropionic acid, compound 7 was N, N-diethyl-S-trityl-L-cysteine). The preparation of compound 7 was as follows: the compound S-trityl-L-cysteine (cas number 2799-07-7) (2.75mmol) was taken and dissolved in 10mL of methanol at 0 ℃ with stirring, acetaldehyde (6.87mmol) was slowly added, reaction was carried out at 0 ℃ for 10min, then sodium borohydride (4.13mmol) was added, and reaction was carried out at 0 ℃ for 2 hours. After the reaction, the methanol is distilled off, and white solid N, N-diethyl-S-trityl-L-cysteine is obtained by silica gel column chromatography.
1H NMR(400MHz,CD3OD)δ7.83(s,1H),7.75–7.68(m,2H),7.54–7.44(m,3H),6.24(dd,J=11.6,2.8Hz,1H),4.96–4.91(m,1H),4.67(dd,J=15.2,4.0Hz,1H),4.49–4.39(m,2H),4.14–4.00(m,1H),3.46–3.37(m,2H),3.28–3.23(m,2H),3.20(dd,J=14.4,3.6Hz,1H),3.10(dd,J=14.8,8.0Hz,1H),2.32(d,J=12.4Hz,1H),2.19–2.09(m,1H),1.87–1.63(m,5H),1.38(t,J=7.6Hz,6H),1.30–1.17(m,4H),1.13–1.03(m,2H);MS(ESI)m/z 455.3[M+H]+.
Compound I-21: the name was (S) -1- (8- (cyclohexylmethyl) -2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) -2-methylpropan-1-one (compound 1(S) -2- (((benzyloxy) carbonyl) amino) -3-cyclohexylpropionic acid and compound 7 was isobutyric acid used).
1H NMR(400MHz,CD3OD)δ7.84(s,1H),7.71(d,J=7.2Hz,2H),7.57–7.40(m,3H),6.22(dd,J=11.6,2.8Hz,1H),4.50(dd,J=15.2,3.6Hz,1H),4.34(dd,J=12.8,3.6Hz,1H),4.28–4.20(m,1H),3.97–3.81(m,1H),3.15–3.05(m,1H),2.17(d,J=12.8Hz,1H),2.09–2.00(m,1H),1.80–1.61(m,5H),1.37–1.22(m,4H),1.20(d,J=6.8Hz,3H),1.16(d,J=6.4Hz,3H),1.09–0.97(m,2H);MS(ESI)m/z 366.3[M+H]+.
Compound I-22: the name 1- ((S) -8- (cyclohexylmethyl) -2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) -2-methylbutan-1-one (compound 1 used was (S) -2- (((benzyloxy) carbonyl) amino) -3-cyclohexylpropionic acid, compound 7 was (S) -2-methylbutanoic acid).
1H NMR(400MHz,CD3OD)δ7.83(s,1H),7.70(d,J=6.8Hz,2H),7.54–7.44(m,3H),6.24(dd,J=11.6,3.2Hz,1H),4.54(dd,J=15.2,4.0Hz,1H),4.35(dd,J=13.2,3.6Hz,1H),4.22–4.14(m,1H),3.97–3.80(m,1H),2.96–2.85(m,1H),2.21(d,J=11.6Hz,1H),2.10–2.00(m,1H),1.80–1.62(m,6H),1.52–1.43(m,1H),1.32–1.22(m,3H),1.20(d,J=7.2Hz,3H),1.14–0.97(m,3H),0.92(t,J=7.2Hz,3H);MS(ESI)m/z 380.3[M+H]+.
Compound I-23: the name is (2S,3R) -2-amino-1- ((S) -8-benzyl-2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) -3-methylpentane-1-one (compound 1 used is N-benzyloxycarbonyl-L-phenylalanine, compound 7 is N-tert-butoxycarbonyl-L-isoleucine);
1H NMR(400MHz,CD3OD):δ8.07(s,1H),7.97–7.86(m,2H),7.50–7.38(m,3H),7.30–7.23(m,3H),7.19(d,J=7.8Hz,2H),5.96(t,J=6.8Hz,1H),4.23–4.13(m,2H),4.06–3.96(m,2H),3.38(d,J=6.8Hz,1H),3.01–2.88(m,2H),1.88–1.77(m,1H),1.16(d,J=7.2Hz,3H),1.10–1.00(m,2H),0.95(t,J=7.6Hz,3H);MS(ESI)m/z 403.2[M+H]+
compound I-24: the name (S) -2-amino-1- ((R) -8- (cyclohexylmethyl) -2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) -3- (4-hydroxyphenyl) propan-1-one (compound 1 used was (R) -2- (((benzyloxy) carbonyl) amino) -3-cyclohexylpropionic acid, compound 7 was Boc-O-tert-butyl-L-tyrosine, 154802-74-1);
1H NMR(400MHz,CD3OD)δ7.78(s,1H),7.72–7.67(m,2H),7.51–7.42(m,3H),7.20(d,J=8.4Hz,2H),6.82(d,J=8.4Hz,2H),6.14(dd,J=11.6,3.2Hz,1H),4.79(dd,J=8.8,6.4Hz,1H),4.30–4.17(m,2H),3.93(dd,J=14.8,3.6Hz,1H),3.47–3.38(m,1H),3.19–3.06(m,2H),2.25(d,J=12.0Hz,1H),1.93–1.85(m,1H),1.81–1.71(m,3H),1.68–1.61(m,2H),1.40–1.34(m,1H),1.31–1.22(m,3H),1.10–1.01(m,2H);MS(ESI)m/z 459.3[M+H]+
compound I-25: the name is (S) -2-amino-1- ((R) -8-isobutyl-2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) -3-mercaptopropane-1-one (compound 1 used is N-benzyloxycarbonyl-D-leucine and compound 7 is N-tert-butoxycarbonyl-S-trityl-L-cysteine);
1H NMR(400MHz,CD3OD)δ7.87(s,1H),7.74–7.68(m,2H),7.55–7.44(m,3H),6.18(dd,J=11.6,2.4Hz,1H),4.77(dd,J=7.2,5.6Hz,1H),4.51–4.42(m,1H),4.37–4.33(m,2H),4.04–3.92(m,1H),3.18(dd,J=14.8,5.6Hz,1H),3.00(dd,J=14.8,7.2Hz,1H),2.21–2.13(m,1H),1.78–1.61(m,2H),1.12(d,J=6.4Hz,3H),0.99(d,J=6.4Hz,3H);MS(ESI)m/z 359.2[M+H]+
compound I-26: the name is (S) -2-amino-1- ((S) -8- ((R) -sec-butyl) -2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) -3-mercaptopropan-1-one (compound 1 used is N-benzyloxycarbonyl-L-isoleucine, compound 7 is N-tert-butoxycarbonyl-S-trityl-L-cysteine);
1H NMR(400MHz,CD3OD)δ7.88(s,1H),7.72(d,J=6.8Hz,2H),7.55–7.45(m,3H),5.81(d,J=7.6Hz,1H),4.76(dd,J=7.2,5.2Hz,1H),4.54–4.42(m,2H),4.40–4.31(m,1H),4.09–3.98(m,1H),3.08(dd,J=14.8,5.2Hz,1H),2.90(dd,J=14.8,7.2Hz,1H),2.27–2.19(m,1H),1.59–1.52(m,1H),1.34–1.26(m,1H),1.15(d,J=6.8Hz,3H),0.96(t,J=7.6Hz,3H);MS(ESI)m/z 359.2[M+H]+.
compound I-27: the name is (S) -2-amino-1- ((S) -8-isopropyl-2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) -3-mercaptopropane-1-one (compound 1 used is N-benzyloxycarbonyl-L-valine, compound 7 is N-tert-butoxycarbonyl-S-trityl-L-cysteine).
1H NMR(400MHz,CD3OD)δ7.88(s,1H),7.73(d,J=7.2Hz,2H),7.54–7.45(m,3H),5.76(d,J=8.0Hz,1H),4.78(dd,J=7.2,5.2Hz,1H),4.56–4.43(m,2H),4.40–4.30(m,1H),4.10–3.98(m,1H),3.09(dd,J=14.8,5.2Hz,1H),2.91(dd,J=14.8,7.2Hz,1H),2.52–2.43(m,1H),1.20(d,J=6.8Hz,3H),1.06(d,J=6.8Hz,3H);MS(ESI)m/z 345.2[M+H]+.
Compound I-28: the name is (S) -2-amino-3-mercapto-1- ((S) -8-methyl-2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) propan-1-one (compound 1 used is N-benzyloxycarbonyl-L-alanine, compound 7 is N-tert-butoxycarbonyl-S-trityl-L-cysteine).
1H NMR(400MHz,CD3OD)δ7.87(s,1H),7.74–7.70(m,2H),7.54–7.44(m,3H),5.91(q,J=6.8Hz,1H),4.74–4.67(m,1H),4.52–4.29(m,3H),4.01–3.87(m,1H),3.09(dd,J=14.8,5.6Hz,1H),2.94(dd,J=14.8,7.2Hz,1H),1.69(d,J=6.8Hz,3H);MS(ESI)m/z 317.1[M+H]+
Compound I-29: the name is (S) -8- (cyclohexylmethyl) -7- (isopropylsulfonyl) -2-phenyl-5, 6,7, 8-tetrahydroimidazo [1,2-a ] piperazine (compound 1 used is (S) -2- (((benzyloxy) carbonyl) amino) -3-cyclohexylpropionic acid, compound 8 is isopropylsulfonyl chloride).
1H NMR(400MHz,CD3OD):δ8.12(s,1H),7.98–7.85(m,2H),7.56–7.43(m,3H),5.99(t,J=6.8Hz,1H),4.62–4.54(m,1H),4.35–4.16(m,2H),3.92–3.85(m,1H),3.32–3.26(m,1H),2.18–2.15(m,1H),2.00–1.97(m,2H),1.66–1.56(m,4H),1.46–0.95(m,6H),1.31(d,J=7.2Hz,6H);MS(ESI)m/z 402.2[M+H]+.
Compound I-30: the name was (S) -2-amino-1- (8- (cyclohexylmethyl) -2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) ethan-1-one (compound 1 used was (S) -2- (((benzyloxy) carbonyl) amino) -3-cyclohexylpropionic acid, compound 7 was N-tert-butoxycarbonylglycine).
1H NMR(400MHz,CD3OD)δ7.84(s,1H),7.74–7.68(m,2H),7.53–7.44(m,3H),6.13(dd,J=11.6,2.8Hz,1H),4.33(d,J=5.6Hz,2H),4.27–4.10(m,3H),3.99–3.86(m,1H),2.25(d,J=12.4Hz,1H),2.12–2.02(m,1H),1.81–1.62(m,5H),1.41–1.32(m,1H),1.31–1.19(m,3H),1.11–1.02(m,2H);MS(ESI)m/z 353.2[M+H]+.
Compound I-31: the title compound was (S) -8- (cyclohexylmethyl) -7- (ethylsulfonyl) -2-phenyl-5, 6,7, 8-tetrahydroimidazo [1,2-a ] piperazine (compound 1 used was (S) -2- (((benzyloxy) carbonyl) amino) -3-cyclohexylpropionic acid, compound 8 was ethylsulfonyl chloride).
1H NMR(400MHz,CD3OD):δ8.07(s,1H),7.96–7.85(m,2H),7.58–7.43(m,3H),5.96(t,J=6.8Hz,1H),4.63–4.53(m,1H),4.35–4.18(m,2H),3.90–3.83(m,1H),3.49–3.36(m,2H),2.16–2.12(m,1H),2.00–1.95(m,2H),1.67–1.56(m,4H),1.46–0.94(m,6H),1.20(t,J=7.2Hz,3H);MS(ESI)m/z 388.2[M+H]+.
Compound I-32: the name is (S) -8-benzyl-7- (isopropylsulfonyl) -2-phenyl-5, 6,7, 8-tetrahydroimidazo [1,2-a ] piperazine (compound 1 used is N-benzyloxycarbonyl-L-phenylalanine, compound 8 is isopropylsulfonyl chloride);
1H NMR(400MHz,CD3OD):δ8.09(s,1H),7.96–7.87(m,2H),7.51–7.39(m,3H),7.31–7.24(m,3H),7.18(d,J=7.8Hz,2H),5.98(t,J=6.8Hz,1H),4.61–4.55(m,1H),4.34–4.16(m,2H),3.92–3.85(m,1H),3.36(d,J=7.8Hz,1H),3.31–3.18(m,1H),3.03(d,J=7.8Hz,1H),1.35(d,J=7.2Hz,6H);MS(ESI)m/z 396.2[M+H]+
compound I-33: the name (S) -2, 6-diamino-1- ((R) -8-isobutyl-2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) hexan-1-one (compound 1 used was N-benzyloxycarbonyl-D-leucine and compound 7 was (S) -2, 6-di-tert-butoxycarbonylaminocaproic acid);
1H NMR(400MHz,CD3OD)δ7.85(s,1H),7.76–7.68(m,2H),7.54–7.43(m,3H),6.18(dd,J=11.6,2.8Hz,1H),4.65(dd,J=8.0,4.8Hz,1H),4.39–4.31(m,3H),4.03–3.94(m,1H),2.97(t,J=8.0Hz,2H),2.22–2.09(m,1H),2.06–1.84(m,2H),1.82–1.68(m,3H),1.65–1.51(m,3H),1.12(d,J=6.4Hz,3H),1.00(d,J=6.8Hz,3H);MS(ESI)m/z 384.3[M+H]+
compound I-34: the compound is named as (S) -1- ((S) -8- (cyclohexylmethyl) -2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) -2-hydroxypropan-1-one (the compound 1 used is (S) -2- (((benzyloxy) carbonyl) amino) -3-cyclohexylpropionic acid, the compound 7 is O-tert-butoxycarbonyl-L-lactic acid, the cas number is 2042339-34-2, and the compound is named as (2S) -2- [ [ (1, 1-dimethyllethoxy) carbonyl ] oxy ] propanoic acid);
1H NMR(400MHz,CD3OD)δ7.82(s,1H),7.74–7.67(m,2H),7.55–7.44(m,3H),6.12(d,J=9.6Hz,1H),4.79–4.71(m,1H),4.64(dd,J=12.8,6.4Hz,1H),4.34–4.24(m,2H),3.91–3.78(m,1H),2.24(d,J=12.0Hz,1H),2.11–2.01(m,1H),1.80–1.63(m,5H),1.42(d,J=6.4Hz,3H),1.36–1.18(m,4H),1.10–0.99(m,2H);MS(ESI)m/z 368.2[M+H]+.
compound I-35: the name (R) -1- (8-isobutyl-2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) -2-methylpropan-1-one (compound 1 used is N-benzyloxycarbonyl-D-leucine and compound 7 is isobutyric acid);
1H NMR(400MHz,CD3OD)δ7.84(s,1H),7.71(d,J=6.8Hz,2H),7.53–7.44(m,3H),6.20(dd,J=11.6,2.4Hz,1H),4.50(dd,J=15.2,3.6Hz,1H),4.35(dd,J=13.2,3.6Hz,1H),4.28–4.21(m,1H),3.97–3.84(m,1H),3.15–3.02(m,1H),2.17–2.06(m,1H),1.75–1.66(m,1H),1.65–1.50(m,1H),1.19(d,J=6.8Hz,3H),1.16(d,J=6.4Hz,3H),1.12(d,J=6.4Hz,3H),0.97(d,J=6.4Hz,3H);MS(ESI)m/z 326.2[M+H]+.
compound I-36: the name (2S,3R) -2-amino-3-methyl-1- ((R) -8-methyl-2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) pentan-1-one (compound 1 used is N-benzyloxycarbonyl-D-alanine and compound 7 is N-tert-butoxycarbonyl-L-isoleucine);
1H NMR(400MHz,CD3OD)δ7.81(s,1H),7.72(d,J=6.8Hz,2H),7.51–7.41(m,3H),6.02(q,J=6.8Hz,1H),4.23–4.13(m,1H),4.06–3.96(m,1H),3.38(d,J=6.8Hz,1H),3.07(dd,J=14.8,5.6Hz,1H),2.95(dd,J=14.8,7.2Hz,1H),1.88–1.77(m,1H),1.48(d,J=7.2Hz,3H),1.16(d,J=7.2Hz,3H),1.10–1.00(m,2H),0.95(t,J=7.6Hz,3H);MS(ESI)m/z 327.2[M+H]+
method for salifying target molecule
The preparation method of the inorganic acid salt comprises the following steps: dissolving a target molecule (1mmol) in 10mL of anhydrous methanol, slowly dropwise adding 5mL of anhydrous methanol solution containing inorganic acid (1mmol) under ice bath, stirring for 30 minutes at the temperature after dropwise adding, and then evaporating methanol at normal temperature to obtain the inorganic acid salt of the target molecule. By the method, hydrochloride (I-10-1), hydrobromide (I-10-2), sulfate (I-10-3) and methanesulfonate (I-10-4) of the compound I-10 are prepared;
the preparation method of the organic acid salt comprises the following steps: dissolving a target molecule (1mmol) in 10mL of anhydrous methanol, slowly dropwise adding 5mL of dry ether solution containing organic acid (1mmol) in ice bath, stirring for 30 minutes at the temperature after dropwise adding, and then evaporating the solvent at normal temperature to obtain the organic acid salt of the target molecule. By this method, maleate salt (I-10-5), succinate salt (I-10-6) and fumarate salt (I-10-7) of compound I-10 were prepared.
Preparation of a mixture of two target molecules
And (3) putting the two target molecules with equal molar weight (1mmol) into anhydrous methanol (5mL), stirring for 10 minutes at room temperature, and evaporating the solvent at room temperature to obtain a mixture of the target molecules. Three target molecule mixtures of (I-3) - (I-10), (I-10) - (I-21) and (I-3) - (I-21) were prepared by this method.
Example 2 evaluation of biological Activity of target molecule
1. Method for testing Gq protein inhibitory activity in vitro
Detection was performed using IP-ONE-Gq KIT (cat # 62 IPAPEEB (1,000 tests)):
1) preparation of IP1 detection reagent
Stimulation Buffer (Stimulation Buffer): 1 volume of 5 XStimB mother liquor was diluted with 4 volumes of distilled water for further use.
Detection Reagent Solutions (Detection Reagent Solutions): 1 volume of 6 Xdetection reagent stock solution was diluted with 5 volumes of Lysis Buffer (lysine & Detection Buffer) for use.
2) Standard curve for IP1
First, IP1 standard was prepared as shown in Table 1 and was used.
TABLE 1 preparation of IP1 Standard
Figure BDA0002794657540000191
② a 384-well plate is taken, 14 mu L of IP1 standard substance is added into each well of the experimental group according to Std7-Std0, 14 mu L of StimB is added into each well of the blank group, and each group has three multiple wells.
③ 3. mu.L of IP1 d2 Reagent solution was added to each well of the experimental group, and 3. mu.L of lysis buffer was added to each well of the blank group.
And fourthly, adding 3 mu L of IP1 Tb Crytate Antibody solution into each hole of the experimental group and the blank group.
And covering the cover, and incubating for 1 hour at room temperature.
And sixthly, detecting by an HTRF reader sample introduction.
And (c) drawing a standard curve of the IP 1.
3) Determination of IC of Gq protein inhibitor50Value of
7 μ L of CHO-M1 cells (which stably express muscarinic receptor 1 in CHO cells (M1) disclosed in the literature "Total Synthesis and Structure-activity relationship study of a series of selective Gq protein inhibitors, Nature chemistry.2016,8, 1035-1041") were added per well according to the Cisbio kit instructions. Each group of three multiple holes.
② 7 microliter StimB is added into each hole of the blank group and the control group, and 7 microliter StimB containing 2 multiplied by the compound to be tested is added into each hole of the experimental group.
③ centrifuging at 600 Xg for 1 minute, mixing well, and incubating at 37 ℃ for 1 hour.
Fourthly, 3 mu L of lysis buffer is added into each hole of the blank group, and 3 mu L of IP1 d2 Reagent solution is added into each hole of the control group and the experimental group.
Fifthly, adding 3 mu L of IP1 Tb Cryptate Antibody solution into each hole.
Sixthly, incubating for 1 hour at room temperature.
And seventhly, detecting the sample injection of the HTRF reader.
The results of analysis are shown in Table 4.
2. Method for testing inhibitory activity of Gi protein in vitro
Detection was performed using CAMP-Gi KIT (cat # 62AM9PEB (1,000 tests)):
1) preparation of cAMP detection reagent
Stimulation Buffer (Stimulation Buffer): 1 volume of 5 XStimB mother liquor was diluted with 4 volumes of distilled water for further use.
Detection Reagent Solutions (Detection Reagent Solutions): 1 volume of the 5 XcAMP Eu Detection reagent mother liquor is taken and diluted with 4 volumes of Lysis Buffer (lysine & Detection Buffer) for use. 1 volume of 5 XcAMP-d 2 antibody stock was diluted with 4 volumes of lysis buffer for use.
2) Preparation of a Standard Curve for cAMP
Preparation of cAMP Standard products as shown in Table 2.
TABLE 2 preparation of cAMP standards
Figure BDA0002794657540000201
② a 384-well plate is taken, 5 mu L cAMP standard substance is added into each hole of the experimental group according to Std7-Std0, 5 mu L StimB is added into each hole of the blank group, and each group has three multiple holes.
③ 5. mu.L of StimB was added to each well of the experimental and blank groups.
And fourthly, adding 5 mu L of cAMP Eu detection reagent solution into each hole of the experimental group and the blank group.
Fifthly, 5 mu L of cAMP-d2 antibody solution is added into each hole of the experimental group, and 5 mu L of lysis buffer solution is added into each hole of the blank group.
Sixthly, the cover is covered, and the mixture is incubated for 1 hour at room temperature.
And seventhly, detecting the sample injection of the HTRF reader.
And drawing a cAMP standard curve.
3) Determination of IC of Gi protein inhibitor50Value of
Add 5. mu.L of a resuspension of CHO-mGlu2 cells (purchased from Girale Biotech, Inc., and CHO cells stably expressing metabotropic glutamate receptor 2) containing 3-isobutyl-1-methylxanthine (IBXM) per well according to Cisbio kit instructions. Each group of three multiple holes.
② 4 microliter StimB is added into each hole of the blank group and the control group, and 4 microliter StimB containing 2.5 multiplied by the compound to be tested is added into each hole of the experimental group.
③ centrifuging at 600 Xg for 1 minute, mixing well, and incubating at 37 ℃ for a suitable time.
Separately, 1 μ L of StimB was added to each well of the blank group, and 1 μ L of forskolin solution with a concentration of 15 μ M was added to each well of the control group and the experimental group.
Centrifuging at 600 Xg for 1 min, mixing, and incubating at 37 deg.C for a proper time.
Sixthly, adding 5 mu L of cAMP Eu detection reagent solution into each hole.
Seventhly, 5 mu L of lysis buffer is added to each hole of the blank group, and 5 mu L of cAMP-d2 antibody solution is added to each hole of the control group and the experimental group.
And incubating at room temperature for 1 hour.
And ninthly, HTRF reader sampling detection.
Analysis of results in r is shown in table 4.
3. In vitro Gs protein inhibitory activity test method
The detection was carried out using CAMP-GS DYNAMIC KIT ((cat # 62AM4PEB (1,000 tests)):
1) preparation of cAMP detection reagent
Stimulation Buffer (Stimulation Buffer): 1 volume of 5 XStimB mother liquor was diluted with 4 volumes of distilled water for further use.
Detection Reagent Solutions (Detection Reagent Solutions): 1 volume of the 5 XcAMP Eu Detection reagent mother liquor is taken and diluted with 4 volumes of Lysis Buffer (lysine & Detection Buffer) for use. 1 volume of 5 XcAMP-d 2 antibody stock was diluted with 4 volumes of lysis buffer for use.
2) Preparation of a Standard Curve for cAMP
First, cAMP standards were prepared as shown in Table 3 and were used.
TABLE 3 preparation of cAMP standards
Figure BDA0002794657540000211
② a 384-well plate is taken, 5 mu L cAMP standard substance is added into each hole of the experimental group according to Std7-Std0, 5 mu L StimB is added into each hole of the blank group, and each group has three multiple holes.
③ 5. mu.L of StimB was added to each well of the experimental and blank groups.
mu.L of cAMP-d2 antibody solution is added to each well of the experimental group, and 5. mu.L of lysis buffer is added to each well of the blank group.
Fifthly, adding 5 mu L of cAMP Eu detection reagent solution into each hole of the experimental group and the blank group.
Sixthly, the cover is covered, and the mixture is incubated for 1 hour at room temperature.
And seventhly, detecting the sample injection of the HTRF reader.
And drawing a cAMP standard curve.
3) Determination of IC of Gs protein inhibitors50Value of
[ 5 μ L of IBXM-containing HEK293- β 2AR cells (a strain of HEK293 cells stably expressing β 2 adrenergic receptors, disclosed in the literature "Total Synthesis and Structure-activity relationship among students of a series of selective Gq protein inhibitors, Nature chemistry.2016,8, 1035-. Each group of three multiple holes.
② 5 microliter StimB is added into each hole of the blank group and the control group, and 5 microliter StimB containing 2 multiplied by the compound to be tested is added into each hole of the experimental group.
③ centrifuging at 600 Xg for 1 minute, mixing well, and incubating at 37 ℃ for a suitable time.
5 mu L of lysis buffer is added into each hole of the blank group, and 5 mu L of cAMP-d2 antibody solution is added into each hole of the control group and the experimental group.
Fifthly, 5 mu L of cAMP Eu detection reagent solution is added into each hole.
Sixthly, centrifuging for 1 minute at 600 Xg, mixing evenly, and incubating for a proper time at room temperature.
And seventhly, detecting the sample injection of the HTRF reader.
The results of analysis are shown in Table 4.
4. Cell growth experiment (CCK-8 detection method)
Cell inoculation: cells in logarithmic growth phase were collected and cell suspension concentration was adjusted to 5X 10 per well3Each cell, 100 μ L per well volume, was seeded into 96-well plates, each set of 4 replicates (marginal wells filled with sterile PBS);
cell culture: after cell attachment, 0% FBS-containing RPMI-1640 medium was starved for 8 hours, and the control group was cultured in 10% (v/v) FBS-containing RPMI-1640 complete medium at 37 ℃ with 5% CO2Continuously culturing in an incubator (culturing for different times according to experimental requirements);
color generation: adding 10 μ L CCK-8 solution (5mg/mL) into the three groups of cells after culturing for 72 hours, terminating the culture within 4 hours, and oscillating on a shaking table at low speed for 10min to fully dissolve crystals;
color comparison: the absorbance (OD) of each well was measured on an ELISA detector, and the absorbance of each well was measured by selecting a wavelength of 450nm, using a cell-free RPMl-1640 culture solution well as a blank group, and using a cell well without stimulation by a compound as a control group. The experiment was repeated three times.
The results are recorded: the inhibition rate of cell growth is ═ x 100% (absorbance value of control group-absorbance value of experimental group)/(absorbance value of control group-absorbance value of blank group) ], and the proliferation rate of cell is ═ x 100% (absorbance value of experimental group-absorbance value of blank group)/(absorbance value of control group-absorbance of blank group) ];
drawing a cell growth curve: the cell growth curve was plotted with time as abscissa and inhibition/proliferation rate as ordinate.
Gra in GraphPad softwareInhibitor concentrations were plotted in phPad Prism mapping software to determine log [ inhibitor [ ]]Variable slope model estimation of IC versus response50
The test results are shown in table 4, and table 4 shows the effect of the obtained compounds on the inhibition of the activity of G protein and the proliferation of antitumor cells.
TABLE 4
Figure BDA0002794657540000221
Figure BDA0002794657540000231
a:IC50: half the effective inhibitory concentration;
b:92.1 (available from Shanghai Rich-balance Biotech, Inc.), MP41 is a typical uveal melanoma cell (available from American ATCC cell Bank), human prostate cancer cell LNCaP (available from American ATCC cell Bank), and human acute promyelocytic leukemia cell HL60 (available from American ATCC cell Bank).
c Stable expression of M1CHO normal cells of the recipient.
The biological activity results show that the molecule has stronger inhibition effect on the G protein, and the effective inhibition concentration IC of most compounds50The value is better than BIM-46174. The results of anti-cell proliferation activity revealed that some of the compounds had very potent inhibitory effects on uveal melanoma cells (92.1 and MP41), with compounds I-3, I-10, I-16 and I-30 also showing unexpected superior activity to BIM-46174. In addition, BIM-46174 has strong toxicity, especially has obvious killing effect on normal cells, and does not have druggability, but the compound in the invention has no obvious killing effect on normal cells, which indicates that the compound has higher safety, indicates that the molecules have the potential of developing novel high-efficiency G protein inhibitors, and has larger potential for treating related tumor diseases, especially uveal melanoma, prostatic cancer, acute myeloid leukemia, pancreatic cancer or glioblastomaThe application value of (2).
The above embodiments are preferred embodiments of the present invention, but the present invention is not limited to the above embodiments, and any other changes, modifications, substitutions, combinations, and simplifications which do not depart from the spirit and principle of the present invention should be construed as equivalents thereof, and all such changes, modifications, substitutions, combinations, and simplifications are intended to be included in the scope of the present invention.

Claims (10)

1. A tetrahydroimidazo [1,2-a ] pyrazine compound or a pharmaceutically acceptable salt thereof, characterized in that: the chemical structure of the tetrahydroimidazo [1,2-a ] pyrazine compound is shown in a formula (I):
Figure FDA0003482988470000011
wherein X is selected from carbonyl; l is selected from-CH-or hydrogen;
R1selected from hydrogen or methyl;
R2selected from cyano groups,
Figure FDA0003482988470000012
Figure FDA0003482988470000013
R3Selected from methyl, amino, hydroxyl,
Figure FDA0003482988470000014
R4Selected from methyl, isopropyl, and isopropyl,
Figure FDA0003482988470000015
2. A tetrahydroimidazo [1,2-a ] pyrazine compound or a pharmaceutically acceptable salt thereof, characterized in that: the tetrahydroimidazo [1,2-a ] pyrazine compound is a compound with the following name:
(I-1) (S) -3-amino-4- ((S) -8- (cyclohexylmethyl) -2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) -4-oxobutanenitrile;
(I-3) (S) -2-amino-1- ((S) -8- (cyclohexylmethyl) -2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) -3- (4-hydroxyphenyl) propan-1-one;
(I-4) (S) -2-amino-1- ((S) -8- (cyclohexylmethyl) -2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) -3- (1H-imidazol-4-yl) propan-1-one;
(I-7) (S) -2-amino-1- ((S) -8- (cyclohexylmethyl) -2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) -3-phenylpropan-1-one;
(I-10) (2S,3R) -2-amino-1- ((S) -8- (cyclohexylmethyl) -2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) -3-methylpentane-1-one;
(I-11) (S) -4-amino-5- ((S) -8- (cyclohexylmethyl) -2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) -5-oxopentanoic acid;
(I-12) (S) -3-amino-4- ((S) -8- (cyclohexylmethyl) -2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) -4-oxobutanamide;
(I-14) (S) -2, 6-diamino-1- ((S) -8- (cyclohexylmethyl) -2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) hexan-1-one;
(I-15) (S) -4-amino-5- ((S) -8- (cyclohexylmethyl) -2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) -5-oxovaleramide;
(I-16) (S) -2-amino-1- ((S) -8- (cyclohexylmethyl) -2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) -3- (1H-indol-3-yl) propan-1-one;
(I-17) (S) -2-amino-3-cyclohexyl-1- ((S) -8- (cyclohexylmethyl) -2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) propan-1-one;
(I-21) (S) -1- (8- (cyclohexylmethyl) -2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) -2-methylpropan-1-one;
(I-22)1- ((S) -8- (cyclohexylmethyl) -2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) -2-methylbutan-1-one;
(I-24) (S) -2-amino-1- ((R) -8- (cyclohexylmethyl) -2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) -3- (4-hydroxyphenyl) propan-1-one;
(I-29) (S) -8- (cyclohexylmethyl) -7- (isopropylsulfonyl) -2-phenyl-5, 6,7, 8-tetrahydroimidazo [1,2-a ] piperazine;
(I-30) (S) -2-amino-1- (8- (cyclohexylmethyl) -2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) ethan-1-one;
(I-31) (S) -8- (cyclohexylmethyl) -7- (ethylsulfonyl) -2-phenyl-5, 6,7, 8-tetrahydroimidazo [1,2-a ] piperazine;
(I-32) (S) -8-benzyl-7- (isopropylsulfonyl) -2-phenyl-5, 6,7, 8-tetrahydroimidazo [1,2-a ] piperazine;
(I-33) (S) -2, 6-diamino-1- ((R) -8-isobutyl-2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) hexan-1-one;
(I-34) (S) -1- ((S) -8- (cyclohexylmethyl) -2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) -2-hydroxypropan-1-one;
(I-35) (R) -1- (8-isobutyl-2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) -2-methylpropan-1-one;
(I-36) (2S,3R) -2-amino-3-methyl-1- ((R) -8-methyl-2-phenyl-5, 6-dihydroimidazo [1,2-a ] piperazin-7 (8H) -yl) pentan-1-one.
3. The tetrahydroimidazo [1,2-a ] pyrazine compound according to claim 1 or 2, or pharmaceutically acceptable salts thereof, characterized in that:
the pharmaceutically acceptable salt is obtained by treating with inorganic acid and organic acid.
4. The tetrahydroimidazo [1,2-a ] pyrazine compound according to claim 3, or pharmaceutically acceptable salts thereof, characterized in that:
the pharmaceutically acceptable salt is sulfate, pyrosulfate, bisulfate, sulfite, bisulfite, phosphate, chloride, bromide, iodide, acetate, caprate, caprylate, acrylate, formate, isobutyrate, hexanoate, heptanoate, propionate, oxalate, malonate, benzoate, chlorobenzoate, methylbenzoate, dinitrobenzoate, hydroxybenzoate, methoxybenzoate, phthalate, phenylacetate, phenylpropionate, phenylbutyrate, citrate, lactate, gamma-hydroxybutyrate, glycolate, tartrate, mandelate and sulfonate.
5. A process for the preparation of tetrahydroimidazo [1,2-a ] pyrazines according to any one of claims 1 to 4, or a pharmaceutically acceptable salt thereof, comprising the steps of:
(1) dissolving a compound 1 with a structural formula shown as a formula 1 and an acid-binding agent A in an organic solvent A, adding a compound 2 with a structural formula shown as a formula 2 for reaction, and removing the organic solvent A after the reaction is finished; adding an organic solvent B for resuspension, adding ammonium acetate, carrying out reflux reaction, and removing the organic solvent B after the reaction is finished; adding water and an extracting agent for extraction, and taking an extracting agent layer; washing, dewatering and concentrating the obtained extractant layer to obtain a compound 3;
Figure FDA0003482988470000021
R4selected from methyl, isopropyl, and isopropyl,
Figure FDA0003482988470000031
(2) Dissolving the compound 3 and an acid-binding agent C in an organic solvent C, adding ethyl bromoacetate, cooling after reaction, and adding water to separate out a compound 4;
(3) dispersing compound 4 and catalyst D in organic solvent D, sealing, and charging H2Reacting, filtering, concentrating the obtained filtrate, and purifying to obtain a compound 5; the catalyst D is Pd/C with the dry content of 10 percent;
(4) dispersing the compound 5 and borane tetrahydrofuran complex solution in an organic solvent E, sealing, introducing argon, carrying out a first reflux reaction, and removing the organic solvent E after TLC monitoring reaction is finished; adding organic solvent F, refluxing for the second time, concentrating, and purifying to obtain compound 6
(5) Preparation of tetrahydroimidazo [1,2-a ] pyrazines by step (a) or by step (B):
(A) dissolving the compound 7 in an organic solvent G, and adding diisopropylcarbodiimide to carry out a first reaction; then adding a compound 6, carrying out a second reaction, and after the TLC monitoring reaction is finished, concentrating and purifying; adding CF to the obtained product under inert atmosphere3COOH and triisopropylsilane, and a third reaction to obtain the tetrahydroimidazo [1,2-a ] according to any one of claims 1 to 4]Pyrazine compounds;
(B) dissolving a compound 8 and triethylamine in an organic solvent H, adding a compound 6, reacting, monitoring by TLC, concentrating and purifying to obtain the tetrahydroimidazo [1,2-a ] pyrazine compound according to any one of claims 1-4;
the compound 7 in the step (A) is N-tert-butyloxycarbonyl-L-asparagine, Boc-O-tert-butyl-L-tyrosine, N-tert-butyloxycarbonyl-N '-trityl-L-histidine, N-tert-butyloxycarbonyl-L-phenylalanine, tert-butyloxycarbonyl-L-aspartic acid-4-tert-butyl ester, tri-tert-butyloxycarbonyl-arginine, N-tert-butyloxycarbonyl-L-isoleucine, N-tert-butyloxycarbonyl-L-glutamic acid-5-tert-butyl ester, tert-butyloxycarbonyl-N-beta-trityl-L-asparagine, (S) -2, 6-di-tert-butyloxycarbonyl-aminocaproic acid, N-tert-butyloxycarbonyl-N' -trityl-L-glutamine, N-tert-butyloxycarbonyl-L-histidine, N-butyloxycarbonyl-L-histidine, N-tert-butyloxycarbonyl-L-histidine, N-butyloxycarbonyl-L-histidine, N-methyl-L-glutamate, N-L-histidine, N-tert-butyloxycarbonyl-L-histidine, N, t-butyloxycarbonyl, N, t-tert-butyloxycarbonyl, N, t-butyloxycarbonyl, N, t-tert-butyloxycarbonyl, N, t-tert-butyloxycarbonyl, N, N-tert-butoxycarbonyl-N' -tert-butoxycarbonyl-L-tryptophan, (S) -2- ((tert-butoxycarbonyl) amino) -3-cyclohexylpropionic acid, isobutyric acid, (S) -2-methylbutyric acid, N-tert-butoxycarbonylglycine, (S) -2, 6-di-tert-butoxycarbonylaminocaproic acid or O-tert-butoxycarbonyl-L-lactic acid;
the compound 8 in the step (B) is isopropyl sulfonyl chloride or ethyl sulfonyl chloride.
6. A pharmaceutical composition having G protein inhibitory activity, characterized by: contains active ingredients or contains active ingredients and a drug carrier; the active ingredient comprises at least one of the tetrahydroimidazo [1,2-a ] pyrazines compound or the pharmaceutically acceptable salt thereof according to any one of claims 1 to 4.
7. The pharmaceutical composition having G protein inhibitory activity according to claim 6, wherein: the active ingredient may also include one or more other pharmaceutically active compounds.
8. Use of the tetrahydroimidazo [1,2-a ] pyrazine compound according to any one of claims 1 to 4 or pharmaceutically acceptable salt thereof, or pharmaceutical composition according to claim 6 or 7 for preparing a G protein inhibitor.
9. Use of the tetrahydroimidazo [1,2-a ] pyrazine compound according to any one of claims 1 to 4 or pharmaceutically acceptable salt thereof and the pharmaceutical composition according to claim 6 or 7 for preparing a medicament for treating tumors.
10. Use according to claim 9, characterized in that: the tumor is melanoma, breast cancer, lung cancer, pancreatic cancer, ovarian cancer, prostatic cancer, gastric cancer, intestinal cancer, head and neck cancer, anal cancer, cancer of the extrahepatic and biliary tract, bladder cancer, bone cancer, brain stem glioma, brain tumor, bronchial adenoma, Burkitt's lymphoma, carcinoid tumor, unknown primary cancer, lymph cancer of the central nervous system, cervical cancer, cancer of children, germ cell tumor, eye cancer, renal cancer, laryngeal carcinoma, liver cancer, non-small cell lung cancer, rectal cancer, salivary gland carcinoma, sarcoma, small intestine cancer, soft tissue sarcoma, uterine sarcoma, testicular cancer, leukemia or blood lymphoma.
CN202011327191.4A 2020-11-24 2020-11-24 Tetrahydroimidazo [1,2-a ] pyrazine compound, composition, preparation method and application thereof Active CN112480122B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202011327191.4A CN112480122B (en) 2020-11-24 2020-11-24 Tetrahydroimidazo [1,2-a ] pyrazine compound, composition, preparation method and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202011327191.4A CN112480122B (en) 2020-11-24 2020-11-24 Tetrahydroimidazo [1,2-a ] pyrazine compound, composition, preparation method and application thereof

Publications (2)

Publication Number Publication Date
CN112480122A CN112480122A (en) 2021-03-12
CN112480122B true CN112480122B (en) 2022-03-22

Family

ID=74933400

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202011327191.4A Active CN112480122B (en) 2020-11-24 2020-11-24 Tetrahydroimidazo [1,2-a ] pyrazine compound, composition, preparation method and application thereof

Country Status (1)

Country Link
CN (1) CN112480122B (en)

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1216545A (en) * 1996-02-16 1999-05-12 生物测量公司 Farnesyl transferase inhibitors

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR2780974B1 (en) * 1998-07-08 2001-09-28 Sod Conseils Rech Applic USE OF IMIDAZOPYRAZINE DERIVATIVES FOR THE PREPARATION OF A MEDICAMENT FOR TREATING CONDITIONS RESULTING FROM THE FORMATION OF HETEROTRIMETER G PROTEIN
EP1233787B8 (en) * 1999-11-09 2005-05-18 Societe De Conseils De Recherches Et D'applications Scientifiques (S.C.R.A.S.) Product inhibiting transduction of g heterotrimeric protein signals combined with another anti-cancer agent for therapeutic use in cancer treatment
FR2803525B1 (en) * 2000-01-06 2002-05-03 Sod Conseils Rech Applic SIGNAL TRANSDUCTION INHIBITOR OF HETEROTRIMERIC PROTEINS ASSOCIATED WITH AN ANTI-HYPERTENSION AGENT IN THE TREATMENT OF ARTERIAL HYPERTENSION

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1216545A (en) * 1996-02-16 1999-05-12 生物测量公司 Farnesyl transferase inhibitors

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
Tetrahydroimidazo[1,2-a]pyrazine Derivatives: Synthesis and Evaluation as Gαq-Protein Ligands;Jim Kuppers,等;《Chemistry—A European Journal》;20200907;第26卷(第55期);第12615-12623页 *

Also Published As

Publication number Publication date
CN112480122A (en) 2021-03-12

Similar Documents

Publication Publication Date Title
CA2995997A1 (en) Compounds and compositions useful for treating disorders related to ntrk
WO2018022761A1 (en) Substituted cyclopentane-amides for treating disorders related to ret
JP2016528197A (en) IDO inhibitor
JP2016519653A (en) IDO inhibitor
WO2018219281A1 (en) 4-pyrimidinediamine small molecular organic compounds and derivatives thereof and use thereof
WO2020234103A1 (en) Identification and use of kras inhibitors
CN110526881B (en) Naphthylamine compound and biologically acceptable salt thereof, and preparation method and application thereof
CN107922348A (en) Bicyclic heterocycle amide derivatives
WO2022135610A1 (en) Tetracyclic compound, pharmaceutical composition thereof and use thereof
CN112480122B (en) Tetrahydroimidazo [1,2-a ] pyrazine compound, composition, preparation method and application thereof
CN110343101A (en) For treating the estrogen receptor degradation agent of breast cancer
Zheng et al. Design, synthesis and biological evaluation of imidazo [1, 5-a] pyrazin-8 (7H)-one derivatives as BRD9 inhibitors
JP7101781B2 (en) Salt morphology as an Akt inhibitor and its crystalline morphology
US20130053434A1 (en) Irreversible inhibitors useful for the treatment of kinase-related pathologies
CN106146525B (en) Three and ring class anaplastic lymphoma kinase inhibitor
Mari et al. 2-Arylmelatonin analogues: Probing the 2-phenyl binding pocket of melatonin MT1 and MT2 receptors
Al-Lakkis-Wehbe et al. Regioselective synthesis of the 1-bromo-4-phenyl-tetrahydro-7-amino-benzocyclohepten-6-one, a subnanomolar aminopeptidase-N/CD13 inhibitor
CN113024460B (en) Tetrahydroisoquinoline compounds as estrogen receptor and histone deacetylase double-target compounds, and synthesis method and application thereof
CN111518095A (en) Azaindole derivatives, and preparation method and application thereof
CN106715445B (en) Big ring N- aryl-tricyclic pyrimidine -2- amine polyethers derivative as FTL3 and JAK inhibitor
WO2021190623A1 (en) Salt and crystalline forms of fgfr4 inhibitor and uses thereof
CN115872996B (en) Estrogen receptor degradation agent compound and preparation method and application thereof
CN115340527B (en) BCL-XL inhibitor and preparation method and application thereof
KR100874209B1 (en) 3,10-dihydro-1h-[1,4]diazepino[5,6-b]indol-2-one derivatives and pharmaceutically acceptable salt thereof, process for preparation thereof and use thereof
WO2023185869A1 (en) Crystalline form of fumarate of pyrroloheterocyclic derivative and preparation method therefor

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant